Language selection

Search

Patent 2493590 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 2493590
(54) English Title: AUTO-STIMULATING CELLS AND METHOD FOR MAKING AND USING THE SAME
(54) French Title: CELLULES AUTOSTIMULANTES ET PROCEDE POUR LEUR PRODUCTION ET LEUR UTILISATION
Status: Dead
Bibliographic Data
(51) International Patent Classification (IPC):
  • C12Q 1/00 (2006.01)
  • A61K 38/00 (2006.01)
  • A61K 47/48 (2006.01)
  • C07K 2/00 (2006.01)
  • C07K 4/00 (2006.01)
  • C07K 5/00 (2006.01)
  • C07K 7/00 (2006.01)
  • C07K 14/00 (2006.01)
  • C07K 14/705 (2006.01)
  • C07K 16/00 (2006.01)
  • C07K 17/00 (2006.01)
  • G01N 33/567 (2006.01)
  • G01N 33/574 (2006.01)
  • A61K 39/00 (2006.01)
  • A61K 35/12 (2006.01)
(72) Inventors :
  • TYKOCINSKI, MARK L. (United States of America)
  • ZHENG, GUOXING (United States of America)
(73) Owners :
  • TRUSTEES OF THE UNIVERSITY OF PENNSYLVANIA (United States of America)
(71) Applicants :
  • TRUSTEES OF THE UNIVERSITY OF PENNSYLVANIA (United States of America)
(74) Agent: RIDOUT & MAYBEE LLP
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2003-07-23
(87) Open to Public Inspection: 2004-02-05
Examination requested: 2008-07-08
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2003/023039
(87) International Publication Number: WO2004/011673
(85) National Entry: 2005-01-25

(30) Application Priority Data:
Application No. Country/Territory Date
10/205,524 United States of America 2002-07-25

Abstracts

English Abstract




Methods for transferring one or more proteins to a cell are disclosed. The
protein or proteins to be transferred are in the form of a fusion protein, and
contain at least one domain encoding for a protein or peptide having trans
signaling and/or adhesion function. The fusion protein is transferred to a
cell by binding to a lipidated protein, which has been incorporated into the
cell membrane. In an additional aspect of the invention, methods of making
fusion proteins having cis signaling capabilities, as well as the ability to
bind with receptors on the cell~s own surface, are provided. Fusion proteins
incorporating GPI or a homing element, and a constimulator or inhibitor domain
can also be directly transferred to the cell surface. Methods for using cells
which have undergone protein transfer according to the present methods are
also disclosed. This includes use in a cancer vaccine, use for treatment of
cancer or autoimmune disease, and use in determening costimulator threshold
levels.


French Abstract

L'invention concerne des procédés pour transférer une ou plusieurs protéines dans une cellule. La ou les protéines à transférer se présentent sous la forme d'une protéine hybride et contiennent au moins un domaine codant pour une protéine ou un peptide présentant une fonction de signalisation et/ou d'adhésion trans. La protéine hybride est transférée dans une cellule par fixation à une protéine lipidée, incorporée dans la membrane cellulaire. Un autre aspect de l'invention concerne des procédés pour produire des protéines hybrides présentant des capacités de signalisation cis ainsi que l'aptitude à se fixer à des récepteurs sur la surface propre de la cellule. Des protéines hybrides, comprenant du GPI ou un élément de <= homing >= et un domaine costimulant ou inhibiteur peuvent également être transférées directement à la surface de la cellule. L'invention concerne également des méthodes pour utiliser des cellules ayant subi un transfert de protéines selon les méthodes de l'invention. Lesdites cellules peuvent être utilisées dans un vaccin contre le cancer, pour le traitement du cancer ou de maladies auto-immunes, et pour déterminer les seuils de costimulants.

Claims

Note: Claims are shown in the official language in which they were submitted.



43

WHAT IS CLAIMED IS:

1. A method for transferring a protein to a cell comprising:
coating the surface of said cell with a first protein, wherein said first
protein is a
lipidated protein; and
contacting said cell with a second protein, wherein said second
protein is a fusion protein having a first domain having affinity for said
lipidated
protein and a second domain capable of binding to a receptor on said cell's
surface.

2. The method of claim 1 wherein said cell is a T cell.

3. The method of claim 2 wherein said T cell is a CD4-positive
T cell.

4. The method of claim 2 wherein said T cell is a CD8-positive
T cell.

5. The method of claim 2 wherein said T cell is a tumor-
infiltrating lymphocyte.

6. The method of claim 2 wherein said T cell has specificity for
a tumor antigen.

7. The method of claim 2 wherein said T cell has specificity for
a viral peptide antigen.

8. The method of claim 1 wherein said cell is selected from the
group consisting of a lymphokine-activated killer cell, a dendritic cell, a
monocyte,
a B cell, a natural killer cell, a neutrophil, an eosinophil, a basophil, a
mast cell, a
keratinocyte, an endothelial cell, an islet cell, a fibroblast, an osteoblast,
a
chondrocyte, a muscle cell, and a neural cell.

9. The method of claim 1 wherein said cell is a stem cell.

10. The method of claim 9 wherein said stem cell is selected from
the group consisting of a hematopoietic stem cell, a mesenchymal stem cell,
and an
embryonic stem cell.

11. The method of claim 1 wherein said second domain of said
fusion protein comprises a costimulator domain that has the ability to
activate said
cell.



44

12. The method of claim 11 wherein said costimulator domain is
selected from the group consisting of B7-1, B7-2, ICAM-1, ICAM-2, ICAM-3,
CD48, LFA-3, CD30 ligand, CD40 ligand, heat stable antigen, B7h, 4-1BB ligand,
OX40 ligand, LIGHT, CD70 and CD24.

13. The method of claim 1 wherein said second domain of said
fusion protein comprises a major histocompatibility complex protein complexed
with
a peptide antigen.

14. The method of claim 1 wherein said second domain of said
fusion protein has the ability to activate a cell selected from the group
consisting of
an antigen-presenting cell, a B cell, a natural killer cell, a neutrophil, an
eosinophil,
a basophil, a mast cell, a stem cell, a keratinocyte, an endothelial cell, an
islet cell,
a fibroblast, an osteoblas, a chondrocyte, a muscle cell, and a neural cell.

15. The method of claim 14 wherein said second domain of said
fusion protein comprises a domain selected from the group consisting of CD40
ligand, TRANCE, Flt-3 ligand, GM-CSF, VEGF, and FGF.

16. The method of claim 1 wherein said second domain of said
fusion protein comprises an inhibitor domain that has the ability to inhibit
said cell.

17. The method of Claim 16 wherein said cell is a stem cell.

18. The method of claim 16 wherein said inhibitor domain has the
ability to inhibit or induce apoptosis in a T cell.

19. The method of claim 18 wherein said inhibitor domain is
selected from the group consisting of Fas ligand and TRAIL and said cell is a
T
cell.

20. The method of claim 1 wherein said second domain of said
fusion protein comprises a poly-histidine tag.

21. The method of claim 1, wherein said fusion protein comprises
a chimeric Fc fusion protein and said lipidated protein is palmitated-protein
A.

22. The method of claim 1 wherein said lipidated protein is
selected from the group consisting of palmitated-protein A and chelator lipid
NTA-
DTDA.



45

23. The method of claim 1, further comprising the step of
purifying said cell prior to coating said surface.

24. The method of Claim 1, wherein more than one second
protein is transferred to said cell.

25. A method of treating a patient for an illness comprising:
coating the surface of a plurality of cells with a first protein, wherein
said first protein is a lipidated protein; and
contacting said plurality of cells with a second protein, wherein said
second protein is a fusion protein comprised of a first domain having affinity
for
said first protein and a second domain capable of binding to a receptor on
said cell's
surface and specific for the treatment of the illness; and
administering an effective amount of said coated cells to said patient.

26. The method of Claim 25, wherein said illness is selected from
the group consisting of cancer, viral infection, autoimmune disease and
alloimmune
disease.

27. The method of claim 25, wherein said cells are injected into
said patient.

28. The method of claim 25, further comprising culturing said
cells ex vivo prior to said adminstration to said patient.

29. The method of claim 25, wherein said cells are lymphokine-
activated killer cells and said patient is a cancer patient.

30. The method of claim 25, wherein said cells are tumor
infiltrating lymphocytes and said patient is a cancer patient.

31. The method of Claim 25, wherein said coating step and said
contacting step take place in vivo.

32. The method of Claim 25, wherein said coating step and said
contacting step take place in vitro.

33. Cells made by the method of Claim 1.

34. The cells of claim 33, wherein said cells are T cells.



46

35. The cells of claim 34, wherein said T cells are CD4-positive T
cells.

36. The cells of claim 34, wherein said T cells are CD8-positive T
cells.

37. The cells of claim 34, wherein said T cells are tumor-
infiltrating lymphocytes.

38. The cells of claim 34, wherein said T cells have specificity
for a tumor antigen.

39. The cells of claim 34, wherein said T cells have specificity for
a viral peptide antigen.

40. The cells of claim 33, wherein said cells are selected from the
group consisting of lymphokine-activated killer cells, dendritic cells,
monocytes, B
cells, natural killer cells, neutrophils, eosinophils, basophils, mast cells,
keratinocytes, endothelial cells, islet cells, fibroblasts, osteoblasts,
chondrocytes,
muscle cells, and neural cells.

41. The cells of claim 33, wherein said cells are stem cell.

42. The cells of claim 41 wherein said stem cells are selected
from the group consisting of hematopoietic stem cells, mesenchymal stem cells,
and
embryonic stem cells.

43. The cells of claim 33 wherein said second domain of said
fusion protein comprises a costimulator domain that has the ability to
activate said
cells.

44. The cells of claim 43 wherein said costimulator domain is
selected from the group consisting of B7-1, B7-2, B7h, ICAM-1, ICAM-2, ICAM-
3, CD48, LFA-3, CD30 ligand, CD40 ligand, heat stable antigen, 4-1BB ligand,
OX40 ligand, LIGHT, CD70 and CD24.

45. The cells of claim 33 wherein said second domain of said
fusion protein comprises a major histocompatibility complex protein complexed
with
a peptide antigen.



47

46. The cells of claim 33 wherein said second domain of said
fusion protein has the ability to activate a cell selected from the group
consisting of
an antigen-presenting cell, a B cell, a natural killer cell, a neutrophil, an
eosinophil,
a basophil, a mast cell, a stem cell, a keratinocyte, an endothelial cell, an
islet cell,
a fibroblast, an osteoblast, a chondrocyte, a muscle cell, and a neural cell.

47. The cells of claim 46 wherein said second domain of said
fusion protein is selected from the group consisting of CD40 ligand, TRANCE,
Flt-
3 ligand, GM-CSF, VEGF, and FGF.

48. The cells of claim 33 wherein said second domain of said
fusion protein comprises an inhibitor domain that has the ability to inhibit
said cell.

49. The cells of Claim 33 wherein said cells are stem cells.

50. The cells of claim 48 wherein said inhibitor domain has the
ability to inhibit or induce apoptosis in a T cell.

51. The cells of claim 50 wherein said inhibitor domain is selected
from the group consisting of Fas ligand and TRAIL and said cells are T cells.

52. The cells of claim 33 wherein said second domain of said
fusion protein comprises a poly-histidine tag.

53. The cells of claim 33, wherein said fusion protein comprises a
chimeric Fc fusion protein and said lipidated protein is palmitated-protein A.

54. The cells of claim 33 wherein said lipidated protein is selected
from the group consisting of palmitated-protein A and chelator lipid NTA-DTDA.

55. The cells of claim 33, wherein said cells are purifed prior to
coating said surface of said cells.

56. The cells of Claim 33, wherein more than one second protein
is transferred to said cell.

57. A pharmaceutical composition for treating a patient for an
illness comprising cells made by the method of Claim 1, in a suitable carrier.



48

58. The composition of Claim 57, wherein said illness is selected
from the group consisting of cancer, viral infection, autoimmune disease and
alloimmune disease.

59 The composition of claim 58, wherein said cells are
lymphokine-activated killer cells and said patient is a cancer patient.

60. The composition of claim 57, wherein said cells are tumor
infiltrating lymphocytes and said patient is a cancer patient.

61. A method for transferring a protein to a cell comprising:
coating the surface of said cell with a fusion protein, said fusion
protein comprising a first domain and a second domain;
said first domain capable of becoming incorporated into a membrane
of said cell, and said second domain capable of binding to a receptor on said
cell's
surface.

62. The method of Claim 61 wherein said first domain comprises
a glycophospholipid.

63. The method of Claim 62, wherein said glycophospholipid is
glycosyl phosphatidylinositol.

64. A method for transferring a protein to a cell of a patient
comprising:
injecting into said patient a fusion protein, said fusion protein
comprising a first domain and a second domain;
said first domain comprising a homing element capable of attaching to
said cell, and said second domain capable of binding to a receptor on said
cell's
surface.

65. The method of Claim 64, wherein said homing element is a
scFv.

66. The method of Claim 64, wherein said homing element is a
cytokine.

67. A cell comprising a cDNA sequence encoding a membrane
protein having the capacity to bind a receptor on the surface of said cell.




49



68. A cell comprising a cDNA sequence encoding a membrane
receptor with the capacity to bind a ligand on the surface of said cell.

Description

Note: Descriptions are shown in the official language in which they were submitted.




CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
_1_
AUTO-STIMULATING CELLS AND METHODS FOR MAKING AND USING
THE SAME
This work was supported in part by Grants R0I CA-74958 and ROl
AI-31044 from the National Institutes of Health.
CROSS-REFERENCE TO RELATED APPLICATION
This application is a continuation-in-part of Application Serial No.
091957,056, filed September 20, 2001, which is a divisional application of
091476,828, filed January 3, 2000, now US Patent No. 6,316,256, expressly
incorporated herein by reference.
FIELD OF THE INVENTION
The present invention relates to novel methods for transferring one or
more proteins to a cell. In addition to other applications, the methodology is
useful
in the treatment of cancer and autoimmune diseases, and for determining
costimulator activation thresholds and cooperative interactions among
costimulators.
The invention further relates to cells painted with extrinsic proteins that
are capable
of triggering receptors on the surfaces of the same cells. Methods of making
such
auto-stimulating cells and using them as therapeutic agents are also
disclosed.
BACKGROUND INFORMATION
T-cells, including cytotoxic T-lymphocytes, are a critical component
of effective human immune responses to tumors, viral infections and other
infectious
diseases. T-ceiis destroy neoplastic or virally infected cells through
recognition of
antigenic peptides presented by MHC class I molecules on the surfaces of
target
cells. Activation of T-cells is dependent upon coordinate signaling through
antigen
receptors and costimulator receptors on T-cell surfaces. Antigen presentation,
in the
absence of simultaneous costimulation, can paradoxically lead to clonal anergy
(See,
Gimmi et al., Proc. Natl. Acad. Sci. USA, 90:6586 (1993)).
Antigen-presenting cells (APC), such as dendritic cells, are geared
towards potent T cell activation, by virtue of their surface MHC and
costimulator
molecules that can trigger their cognate receptors on specific T cells and
thereby
provide both critical signals to them (See, Watts et al., Curr. Opin.
Immunol.,



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
2
11:286 (1999); Freedman et al., Cell. Tmmunol., x37:429 (1991)). Thus,
according
to the prevalent APC-centric view of T cell regulation, T cell activation is
generally
'viewed as being under the control of professional APC, via intercellular
"t~atas"
signaling.
Many mechanisms contribute to the escape of tumor cells and virally
infected cells from immune surveillance. One of the mechanisms is that these
cells
lack the costimulatory molecules required for T-cell activation. "Active"
immunotherapeutic strategies have been developed that are predicated upon
expressing costimulators on tumor cell, and other antigen-presenting cell,
surfaces.
An alternative "passive" immunotherapeutic approach involves the steps of
recovering tumor-infiltrating lymphocytes (TIL) from tumor beds, or T cells
from
the blood of cancer patients, expanding the numbers of these T cells ex viuo
with
lymphokines, and injecting them back into the patient.
A major limiting factor for the clinical application of therapeutic T
cells is their loss of activity once injected into patients. This is generally
believed to
be a consequence of a deprivation of T cell activators, such as soluble
cytokines and
surface costimulators. Costimulation is required not only for the early
activation of
T cells, but also for the later maintenance of their post-activation effector
capacity,
referred to as "effector costimulation." In an effort to maintain therapeutic
T cells
2G in a fully activated state, activating cytokines, such as IL-2, have been
administered
systemically to patients receiving therapeutic T cells, albeit with IL-2
toxicities and
insufficient therapeutic benefit.
T cells also bear inhibitory receptors. The fate of T cells following T
cell receptor (TCR) stimulation is guided by the integration of costimulatory
and
inhibitory receptor inputs. As indicated, costimulatory ligands on APCs
trigger
cognate receptors on T cells, with resultant enhancement of T cell
proliferation,
cytokine secretion, and differentiation. In contrast, binding of inhibitory
ligand
molecules on various cells to cognate receptors or. responding T cells
diminishes
effector functioning, by inducing T cell unresponsiveness or apoptosis.



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
3
Professional antigen-presenting cells (APC), by virtue of the surface
costimulatory molecules, are geared towards potent T-cell activation. APC can
be
converted into deletional APC, or "artificial veto cells", by expressing
coinhibitors
at their surfaces. This is discussed, for example, in U.S. Patent Nos.
5,242,687;
5,601,828; and 5,623,056. Such coinhibitors bind to coinhibitor receptors on
cells,
leading to T-cell inactivation.
One approach for expressing costimulators and coinhibitors on APC,
such as tumor cells, is gene transfer. When used for APC and tumor cell
engineering, gene transfer techniques have shortcomings. Fox example, APCs,
including tumor cells, are often poorly transfectable. In addition,
transfection
proceedings are cumbersome and time-consuming. Furthermore, expressing more
than a costimulator (oi- coinhibitor) is difficult. These and other issues
have
impeded the widespread application of gene therapy for APC and tumor cell
engineering.
Protein transfer offers a number of advantages over gene transfer for
engineering APCs and other cells. These advantages include the ability to
modify
poorly transfectable cells (for example, biopsy-derived tumor cells), the
simplicity
of expressing multiple proteins on the same cell surface, and the relative
ease and
rapidity of the procedure. The successful use of recombinant GPI-modified
costimulator and MHC protein derivatives for protein transfer has been
reported.
(See, Brunschwig, et aI. J. Irn»aunol. , 155:5498 (1995); McHugh, et al; Proc.
Natl.
Acad. Sci. USA, 92:8059 (1995); and McHugh, et al. Ca»cer Res., 59:2433
(1999)). A shortcoming of the GPI protein transfer strategy, however, resides
in
scaling up the purification of GPI proteins from membranes of transfected
cells. The
successful use of protein conjugates consisting of recombinant Fc-modified
costimulator derivatives complexed to palmitated protein A for protein
transfer has
also been reported (See, U.S. Pat. No. 6,316,256; Chen, et al. J. Immunol.,
164:705 (2000); ~heng, et al. Cancer Res., 61:8127 (2001)). Yet another
protein
transfer method entails appending hiss-tagged costimulators to cells pre-
treated with
the chelator lipid, NTA-DTDA, whose NTA groups bind the hexahistidine tags
(See,
van Broekhoven et al. J. Immunol., 164:2433 (2000)).



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
4
Kim and Peacock, J. Immunol. Methods, 158:57 (1993), report the
use of palmitate-conjugated protein A for coating cells with artificial
receptors
which facilitate intercellular interactions. More specifically, a method is
reported
for attaching an antibody onto the surface of a cell using palmitated protein
A. The
article does not teach use of a lipidated protein for attachment of anything
other than
an antibody to a cell. As such, their modified cells serve only as artificial
receptors
for antigens.
Phillips et al., Immufiity, 5:163-172 (August, 1996) report the
preparation of a fusion protein using a CD8 leader segment, the Fc domain, of
immunoglobin and the ectodomain of a type II membrane protein, CD94. The
present transfer methods are applicable to both type I and type II proteins
and are
neither taught nor suggested in the article.
Darling, et al., Ge~ze Therapy, 4(12):1350-60 (Dec. 1997) report the
use of a biotin/avidin-based system for protein transfer. This method involves
biotinylation of the target cell, attachment of an avidin group to the protein
to be
transferred, and combining the biotinylated target cell and the avidin-tagged
protein.
This method has significant limitations, including its dependence on covalent
modifications that could perturb multiple proteins on cell surfaces.
Certain T cell costimulators, including B7-1, B7-2, 4-1BB ligand, and
OX40 Iigand, are expressed on T cells themselves, either under normal
conditions
or in diseased states (See, Carreno et al. Annu. Rev. Immunol., 20:29 (2002);
Nakamura et al. J. Exp. Med., 194:629 (2001); Kochli et al. Immunol. Lett.,
65:197 (I999); Wolthers et al. Eur. J. ImmunoL, 26:1700 (1996); Takasaki et
al.
Intern. Med., 38:175 (1999); Weintraub et al. J. Immunol., 159:4117 (1997);
2S Weintraub et al. Clin. Immunol., 91:302 (1999)). In addition, it is known
that T
cells can acquire costimulators from APC via intercellular transfer (See,
Hwang et
al. J. Exp. Med., 191:1137 (2000); Sabzevari et al. J. Immunol., 166:2505
(2001)),
just as they can acquire MHC:peptide antigen complexes via intercellular
transfer
(See, Lorber et aI. J. Immunol., 128:2798 (1982); Hudrisier et al. J.
Immunol.,
166:3645 (2001)). These costimulators and MHC:peptide antigen complexes on T



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
cells have been presumed to trigger cognate receptors on neighboring T cells
in
traps.
After T cell triggering, first the inhibitory receptor Fas, and then its
cognate ligand, Fas ligand, are sequentially upregulated on T cell surfaces.
While
5 some have surmised, on the basis of indirect evidence, the possibility that
T cell
"suicide" (as a concomitant of T cell "fratricide/sororicide") might result
from such
Fas ligand:Fas pairing at the cell surface (See, Brunner et al. Nature,
373:441 (1995);
Dhein et al. Nature, 373:438 (1995); Ju et al. Nature, 373:444 (1995)),
definitive
proof that this mechanism is indeed operative (for example, enforced
expression of
the ligand:receptor pair) is lacking. Furthermore, while there are other
examples of
ligand:receptor pairs that are naturally co-expressed on T cell surfaces, for
example,
CD58 (LFA-3) and its cognate receptor CD2 (See, Springer et al. Annu. Rev.
Immunol., 5:223 (1987)), the functional implications of this pairing at the
same cell
surface (fox example, the potential for continuous triggering and/or
competitive
IS blockade of incoming tf~ahs signals) have been ignored.
The concept of "autocrine signaling", wherein a cell secretes a
soluble protein ligand that binds and signals through one of its own native
receptors,
has been discussed in the prior literature (See, e.g., Hoffbrand A.V., Semin.
Hef7zatol., 30:306 (I993). For example, certain leukemia cells secrete soluble
growth factors that can bind to the cell's own receptors, prompting the notion
that
autocrine signaling results from this ligand:receptor interaction and may play
a role
in leukemogenesis. However, this literature has dealt exclusively with soluble
ligands which bind to the cell's receptors. No therapeutic cells in the art
have been
designed with membrane-embedded proteins with the capacity to trigger their
own
activating or inhibitory receptors.
There remains a need, therefore, for methods of efficient and
quantitative transfer of proteins and peptides to cells. A further need is to
provide
such methods in which immunoregulatory or other molecules that retain their
function can be attached to cells of interest, including membrane-binding
proteins
that enable cells to auto-stimulate themselves.



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
6
SUMMARY OF THE INVENTION
The present invention has met the above needs, by providing methods
for quantitative transfer of a domain having traps-signaling and/or adhesion
function
onto a cell surface. Typically, the domain will be the extracellular domain
having
one or both of these functions. In a preferred embodiment, the extracellular
domain
of an immuno-regulatory molecule is used. More specifically, the present
methods
provide a two-step protein transfer approach, which permits delivery of graded
amounts of proteins to a cell surface. The methods utilize a fusion protein
comprised of at least two domains, one of which preferably encodes a molecule
having immunoregulatory function. By adding the fusion protein to cells coated
with a lipidated protein, fine titration of, for example, the immunoregulatory
molecule's extracellular domain is achieved.
The present protein transfer methods have wide application. For
example, the methods have been used to establish that costimulator thresholds
exist,
and that the levels of surface costimulator on APC can dictate both the
magnitude
and the quality of evoked T-cell responses. The present methods are also
applicable
to the generation of cancer vaccines; these vaccines show significant anti-
tumor
effects ira vivo. Furthermore, the methods can be used to generate artificial
veto
cells, expressing one or more coinhibitors, that can be used to delete
pathogenic T-
cells. Cells produced according to the present methods are therefore useful in
the
treatment of cancer and also in the treatment of autoimmune diseases. The
methodologies described herein can also be used in the establishment of animal
models and for the study of immunological issues regarding, for example, T-
cell
activation, use of costimulators to override apoptotic signals, function of
coinhibitors versus costimulators, synergy of costimulators used in the
treatment of
cancer, and use of coinhibitors in the treatment of autoimmune diseases.
In an additional aspect, the present invention provides a method of
producing therapeutic cells coated with one or more extrinsic protein ligands
that
can not only trigger cognate receptors on neighboring cells (tratas
signaling), but
also stimulate themselves via interaction of the ligand with a receptor on the
cell's



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
7
own surface (cis signaling). As used herein, "cis signaling" refers to the
receptor:ligand interaction on the surface of a cell that provides signals to
the
interior of that cell, such signals triggering protein phosphorylation or
dephosphorylation, DNA transcription or other cellular events. As described
above,
such cells have wide application and can be used in the treatment of many
diseases,
including cancer and autoimmune diseases. The fusion proteins can be coated on
the
cell surface in a two step method, as described above, or can alternatively be
applied in a one-step method, by painting a fusion protein having a domain of
a
molecule, such as a glycophospholipid, which is capable of incorporating into
the
cell membrane directly. Such a fusion protein would have, as a second domain,
the
signaling protein of interest.
It has been discovered that when ligands of interest are immobilized
on the cell surface, surprisingly, they are still capable of binding with the
cognate
receptor on the same cell and providing the stimulation or inhibition
necessary to
activate (or turn offj the cells. This was not expected, based on the prior
art.
Moreover, it has been found that the enforced ligand:cognate interaction can
give a
much stronger interaction than that found with intercellular traps signaling.
For example, in the case of T cells of the immune system, use of an
activating protein renders the therapeutic T cells suitable for use in the
treatment of
cancer and virally mediated diseases, whereas use of an inhibitory protein
renders
the therapeutic cells suitable to function as regulatory T cells for treatment
of
autoimmune and alloimmune disorders. Therapeutic auto-stimulating cells,
corresponding to numerous other immune and non-immune cell types (including
various stem and progenitor cells) can be similarly configured.
The present invention further provides methods for making the
present therapeutic auto-stimulating cells, and methods for using these cells
in the
study and treatment of cancer, viral, autoimm~une and alloimmune diseases, as
well
as any one of a number of conditions in which auto-stimulating cells may be
beneficial. Other methods for studying and treating these diseases and
conditions,
both in vivo and ex vivo, are also disclosed.



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
8
It is therefore an aspect of the invention to provide novel therapeutic
cells that have the capability of auto-stimulating themselves.
Another aspect of the present invention is to provide methods for
producing such auto-costimulating cells, with protein transfer being a
preferred
method for achieving this end.
Yet another aspect of the present invention is to provide therapeutic
cells for the study and treatment of cancer, viral, autoimmune and alloimmune
diseases and disorders, as well as any one of a number of conditions in which
auto-
stimulating cells may be beneficial.
These and other aspects of the invention will be apparent based upon
the following description and appended claims.
BRIEF DESCRIPTION OF THE FIGURES
Figure I, including IA, IB and 1C, demonstrates the efficacy of
coating cells with a lipidated protein, according to the methods of Example 1.
IS Figure 2 provides the SDS-PAGE analysis of recombinant B7-I-Fcy~
prepared as described in Example I.
Figure 3 demonstrates that the present methods achieve fusion protein
transfer (Fig. 3A), in a duantitative manner (Fig. 3B), as described
in.ExampIe I.
Figure 4 demonstrates the stimulation of T-cell proliferation in the
presence of various proteins (as indicated) using either PHA (Fig. 4A) or anti-
CD3
mAb (Fig. 4B) as a first signal, as described in Example 1.
Figure 5 demonstrates B7-I threshold concentrations for T-cell
proliferation using either PHA (Fig. SA) or anti-CD3 mAb (Fig. 5B) as a first
signal, as described in Example 1.
Figure 6 provides a comparison of B7 concentration thresholds for
IFN-y versus IL-2 pzoduction using either PHA (Fig. 6A) or anti-CD3 mAb (Fig.
6B) as a first signal, as described in Example I.
Figure 7 provides comparative single-cell analyses of B7-1
concentration thresholds for IFN-y versus IL-2, as described in Example 1.



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
9
Figure 8 shows the effect of reaction temperature during protein
transfer on the stability of transferred protein, as described in Example 2.
Figure 9 demonstrates the efficacy of the present cancer vaccines in
protecting a patient against a post-immunization tumor challenge, as described
in
Example 3.
Figure 10 demonstrates the efficacy of the present cancer vaccines in
treating a pre-existing tumor, as described in Example 4.
Figure 11 demonstrates the efficacy of the present cancer vaccines by
intratumoral injection of costimulators, as described in Example 5.
Figure 12 shows the results of the JAM assay, as described in
Example 6.
Figure 13, including I3A and 13B, demonstrates the efficacy of
painting both CD4''~ and CD8+ murine T cells with two different costimulators,
and
the enhanced T cell proliferative potential of these costimulator-painted T
cells,
according to the methods of Example 7.
Figure 14, including 14A and 14B, demonstrates the enhanced
proliferative potential of a murine T cell line (EL-4) and purified human T
cells
after costimulator painting, according to the methods of Example 7.
Figure I5 documents the CD28-dependence of the enhanced
proliferation observed fer T cells painted with the B7-1 costimulator,
according to
the methods of Example 7.
Figure 16, including 16A and 16B, demonstrates that costimulator
painting enhances the proliferative response of TCR-transgenic T cells to
their
specific cognate peptide antigen, according to the methods of Example 8.
Figure 17, including 17A, 17B and 17C, demonstrates via cell-
mixing experiments (that employ mitomycin C-inhibited cells) that auto-
costimulation is a major contributor to the enhanced proliferation of
costimulator-
painted T cells, according to the methods of Example 9.
Figure 18, including 18A and 18B, demonstrates via cell-mixing
experiments (that employ CFSE-labeled cells) that that auto-costimulation is a
major



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
contributor to the enhanced proliferation of costimulator-painted T cells,
according
to the methods of Example 9.
Figure 19 demonstrates that the relative contribution of cis auto-
costimulation, as opposed to intercellular traps costimulation, in a culture
of
5 costimulator-painted T cells increases when the cells are diluted, according
to the
methods of Example 10.
DETAILED DESCRIPTION OF THE INVENTION
The present invention is directed to methods for transferring one or
10 more proteins to a cell, generally comprising the steps of coating the
surface of a
cell with a first protein, which is a lipidated protein, and contacting the
coated cell
with a second protein, which is a fusion protein. The fusion protein is
comprised of
a first domain having affinity for the lipidated protein and a second domain
of a
peptide, protein, or derivative or fragment thereof, having tfaoZS signaling
and/or
adhesion function. Preferably, the extracellular domain is used, and the
second
domain has immunoregulatory function. "Derivative", as used in reference to
peptides and proteins, refers to variants of peptides and proteins such as
analogues
wherein, for example, one or more amino acids within the peptide chain has
been
deleted, added or replaced with an alternative amino acid. A "fragment" refers
to a
portion of the amino acid sequence of a peptide or protein. It will be
understood
that "derivatives" and o fragments" of peptides and proteins retain the
physiological
function of the wild type peptide or protein and thus are biologically active.
The present methods are applicable to any cell having a lipid bilayer
membrane. For example, any kind of a patient's autologous cells can be used,
harvested by any means known in the art. Use of any allogeneic mammalian cell
line is also within the scope of the present invention. Examples of allogeneic
cell
lines suitable for use in the present invention include, but are not limited
to, EL-4
cells (mouse thymoma cells), 293 cells (human kidney cells), I~562 cells
(human
leukemia cells), Daudi cells (human B cell line) and 3'Y cells (human B cell
line).
These cells are commercially available from the American Type Culture
Collection,



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
11
Manassas, VA. Non-commercially available cell lines are also within the scope
of
the present invention. Use of isogeneic and xenogeneic cells is also within
the scope
of the present invention.
Any protein that can be lipidated is suitable for use in the present
methods. Examples include, but are not limited to, protein A and protein G,
both of
which are commercially available. Similarly, any lipid can be used to prepare
the
lipidated protein. Lipids having carbon chains between about 12 and 22 are
preferred, with a carbon chain of 16 (palmitate) being most preferred. The
length
of the lipid chain can be varied based upon the needs and desires of the user.
It will
be understood to those skilled in the art that the lipidated portion of the
first protein
will become attached to or incorporated into the phospholipid bilayer that
makes up
the membrane of the cell, and that this is what is meant by the phrase
"coating the
surface of the cell" as that phrase is used herein.
The amount of lipidated protein used to coat the cell may also vary
based on the needs and desires of the user, and based on the particular lipid
and
particular protein selected. Preferably, enough lipidated protein is used to
coat the
entire cell. This amount will typically be at least about 30 micrograms of
lipidated
protein for every 5 x 106 cells.
Following coating of the cells, the cells are then contacted with a
second protein. The second protein is a fusion protein in which two different
domains have been fused, such as through recombinant DNA technology standardly
used in the art, to create a single DNA sequence. The first domain can be
attached
at either the amino terminus or the carboxyl terminus of the fusion protein.
The
first domain encodes a peptide, protein, or derivative or fragment thereof,
what has
affinity for the lipidated protein. Thus, the protein used in the lipidated
protein is
ideally selected in conjunction with the protein encoded by the first domain
of the
fusion protein, so that proteins having affinity for one another are used.
Affinity
between the proteins can be determined by Biacore technology or other methods
familiar in the art. Because of this affinity, the fusion protein binds to the
lipidated
protein, which has already been incorporated into the cell membrane. In this



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
12
manner, the fusion protein is transferred to the cell. A particularly
preferred
combination uses a palmitated protein A and a first domain encoding an Fc
region.
For example, the Fc region of human immunoglobulin GI (IgGI), designated Fcyl,
can be used. Other suitable first domains in the fusion protein include
leucine zipper
protein domains and single-chain Fv derivative domains.
The second domain of the fusion protein encodes a peptide, protein,
or derivative or fragment thereof, having immunoregulatory function and
capable of
tYarcs signaling to a second cell. Examples include, but are not limited to,
costimulators and coinhibitors. Any suitable costimulator can be used
including but
IO not limited to B7-I, B7-2, CD48, ICAM-I, ICAM-2, ICAM-3, LFA-3, CD30
Ligand (CD30L), CD40 Ligand (CD40L), 4-IBB Ligand (4-1BBL), OX40 ligand,
CD70, CD24, and heat stable antigen. Similarly, any suitable coinhibitor can
be
used including, but not limited to, CDB, Fast, PP14 and TRAIL.
Significantly, the fusion protein of the present invention can be either
IS a type I or type II protein. Methods for transferring a type II protein to
a cell have
never been reported. Because the methods of the present invention are equally
applicable to type T and type II proteins, they provide a significant advance
over the
art. Examples of type I membrane proteins include B7-1, B7-2 and CD48;
examples of type II membrane proteins include Fas ligand (Fast or CD95L),
20 CD40L, and 4-IBBL. For the type TI proteins, the first domains are fused at
the
carboxyl termini of the type II proteins in order to preserve the functional
ends of
the molecules. These lists are not exhaustive of the costimulators,
coinhibitors and
other proteins that can be transferred according to the present invention; the
lists
reflect all forms of the various molecules including, but not limited to,
human and
25 murine forms.
Another significant advance provided by the present methods is that
following transfer of the fusion protein to the cell, the portion of the
fusion protein
having trajiS signaling function retains this function. Thus, the cells
prepared
according to the present methods are capable of eliciting an immune response
by
30 binding to, and tt~ans signaling through a counter-receptor on a second
cell.



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
13
In addition, more than one fusion protein can be used to coat a single
cell. In this manner, two, three, four, or more traps signaling, fox example,
immunoregulatory proteins, can be transferred to a cell. In the case of
costimulators
and coinhibitors, combinations of such proteins can be chosen to have the
greatest
irnmunological effect; combinations having additive or even synergistic
benefits can
be selected and used according to the present methods.
The present methods are further unique in that proteins can be
delivered to a cell's surface in a quantitative manner. As noted above, it is
preferred to use enough lipidated protein to fully coat the cell. The amount
of
fusion protein that becomes transferred to the cell is therefore determined by
the
amount of fusion protein used to contact the coated cell; thus the amount of
fusion
protein is the limiting or determinative factor. When using more than one
fusion
protein, predetermined ratios of fusion proteins can be used to contact the
coated
cell; protein will be transferred to the cell in these approximate ratios.
The present methods can be effected either in vivo or in vitro. In ift
vivo methods, the lipidated protein and the fusion protein are injected
directly into a
patient. The injection can occur sequentially (with the lipidated protein
first) or
concurrently, with premixing of the lipidated protein and the fusion
protein(s).
Injection can be localized, for example, intra-tumoral, or systemic, for
example,
into a vessel. The present methods, and cells produced thereby; in contrast to
other '
art-reported methods, have particularly high protein stability, making the
present
method practical for in vivo application. In vitro methods involve the
extraction of
cells from a patient, and the subsequent coating and contacting of the cells;
alternatively, commercially obtained allogeneic cells can be used. In either
case, the
treated cells can then be injected into a patient.
All of the above descriptions relating to cell type, first proteins,
second proteins, and in vivo, ira vitro and other delivery techniques apply
equally to
all embodiments of the invention disclosed herein.
1'he present invention is further directed to methods for determining
costimulator activation thresholds in T-cells. These methods generally
comprise



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
14
transferring one or more fusion proteins to a cell, in the manner described
above.
The cells to which protein has been transferred are then mixed with T-cells. T-
cells
can be, for example, harvested from peripheral blood mononuclear cells by
methods
known in the art. T-cell proliferation, if any, can be measured, as can
cytokine
secretion levels according to means known in the art, such as those described
in the
Example section.
The present invention is further directed to methods for treating an
illness using the present protein transfer technology. These methods generally
comprise administering to a patient an effective amount of the cells prepared
ih vitro
according to the method described above, or administering the proteins in
vivo. The
method can be performed by either in vivo or in vitro protein transfer of the
fusion
proteins) to the target cells. Fox ih vitro methods, either extracted
autologous cells
or allogeneic cells are coated with a lipidated protein and contacted with one
or
more fusion proteins. An effective amount of these cells are then administered
to a
patient. For in vivo methods, lipidated protein and one or more fusion
proteins are
administered to a patient in an amount sufficient to result in transfer of an
effective
amount of fusion proteins) to an effective amount of cells.
"Illness" as used herein refers to cancer, viral, and autoimmune and
alloimmune diseases, including but not limited to melanoma, ovarian cancer,
breast
cancer, colon cancer, pancreatic cancer, hepatic cancer, lung cancer,
glioblastoma
multiforme, prostate cancer and leukemia; viral infections, such as chronic
viral
infections with HBV, HCV, HTLV-l, HTLV-II, EBV, CMV, HSV-I, HSV-II, and
KSHV; arthritis, rheumatoid arthritis, asthma, graft-versus-host disease,
organ
rejection, systemic lupus erythematosis, atopic allergy, inflammatory bowel
disease,
multiple sclerosis and allergic dermatitis. The methods are particularly
applicable to
treatment of cancer, in that the lipidated protein and fusion proteins) in the
ifi vivo
methodology, or the coated and contacted cells in the it vitro methodology,
can be
directly injected into one or more tumors of the patient. "Patient" is used
herein to
refer to members of the animal kingdom, including humans. The present methods



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
are generally applicable to patients capable of generating at least a minimal
immune
response.
An effective amount of cells produced by the present protein transfer
methods should be used in the present treatment methods. The effective amount
is
5 that amount of cells that will deliver .the amount of protein to a patient
needed to
bring about a desired result. Generally, the desired result can be, for
example,
stimulation of an immune response or suppression of an immune response. In the
case of cancer treatment, an effective amount would be that amount which would
protect a patient against tumor growth or reduction, if not elimination, of
tumors.
10 In the case of autoimmune or alloimmune disease, an effective amount would
be that
amount which would alleviate if not eliminate one or more symptoms of the
autoimmune or alloimmune disease being treated. In the case of treatment of
viral
diseases, an effective amount would be that amount which would induce a
reduction,
if not elimination, of the viral infection. It will be understood that the
effective
15 amount will vary from patient to patient depending on such factors as the
patient's
size, the condition of the patient's immune system, the patient's ability to
mount an
immune response, and the type and severity of the illness. The appropriate
effective
amount for each patient can be determined by one skilled in the art, and will
generally be at Ieast about 10' modified cells or 100 p,g Fc fusion protein
intratumorally. In the case of auto-stimulated T cells, described more fully
below,
the effective amount will generally be at least about 108 modified cells. The
present invention is further directed to a cancer vaccine comprising cells
prepared
according to the present protein transfer methods contained in a suitable
carrier.
Any suitable carrier can be used, provided compatibility problems do not
arise.
ZS Examples include PBS, and serum-free medium. The vaccine can include a
variety
of fusion proteins; different cells each having a different fusion protein, or
cells
having more than one fusion protein attached thereto, can be used for example.
Thus, a "cocktail" of immunoregulatory proteins can be contained in the
present
vaccines, and can be introduced to a patient according to the present methods.
The
particular immunoregulatory proteins to use in a cocktail can be determined by
one



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
16
skilled in the art based upon such factors as the patient being treated and
the type
and severity of the patient's illness. Different combinations could be used to
treat
different types of tumors. The cocktail can be pre-mixed and injected into a
tumor
bed, thereby leading to tumor suppression. The vaccines have been found
effective
in both pre-immunizing recipients against a subsequent tumor challenge and in
the
treatment of established tumors.
The present invention is further directed to methods for enhancing the
efficacy of therapeutic cells by transferring proteins to the surface of cells
to provide
auto-stimulating ability. The method generally comprises coating the surface
of a
cell with a first protein, the first protein being a lipidated protein, and
contacting
the cell with a second protein. The second protein is a fusion protein
comprised of a
first domain having affinity for the first protein and a second domain having
cis
signaling function, and is capable of binding to a receptor on the cell's own
surface.
In an alternative one-step embodiment, also preferred, the method of
transfer comprises coating the surface of a cell with a fusion protein having
a first
domain and a second domain, the first domain capable of becoming incorporated
into a membrane of a cell, and the second domain capable of binding to a
receptor
on the cell's own surface. The first domain of the fusion protein of the one-
step
method will preferably by a glycophospholipid, more preferably glycosyl
phosphatidylinositol (API). The second domain will comprise the protein of
interest, such as the costimulators and inhibitors described above.
The fusion protein transferred to the surface of the cell is a ligand
that binds to a receptor on the same cell, and in so doing, engenders a change
in the
physiological state of the cell. "Auto-stimulating cell" or "auto-stimulating
ability" as used herein refers to a cell bearing on its surface an extrinsic
ligand,
generally a protein, capable of engaging and triggering a receptor on its own
surface. Such engagement of the ligand:receptor on the cell surface results in
a cis
signal as that term is defined above; such a signal will include stimulatory
signals
and inhibitory signals, as those terms are understood in the art. Thus the
terms
"auto-stimulating" and "cis signaling" refer to both stimulatory and
inhibitory



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
17
outcomes from the receptor:ligand interaction. The present invention stems
from the
discovery that if one paints a ligand onto cognate receptor-bearing cells,
receptor
triggering can ensue.
Preferred embodiments of the present invention are within the
immunotherapeutic realm. In a preferred embodiment, the present invention
comprises the painting of a patient's autologous T cells with proteins, for
purposes
of maintaining these cells in an activated state after they are injected back
into a
patient. This is especially useful for enhancing the activity of therapeutic T
cells
with anti-tumor or anti-viral properties. Fox purposes of activating T cells
in this
context, any suitable costimulator can be used including, but not limited to,
B7
family costimulators (such as B7-l, B7-2, B7-h/B7rp-1), 4-1BB ligand, OX40
ligand, LIGHT, CD70, CD24, CD4~, ICAM-1, ICAM-2, ICAM-3, LFA-3, and
CD30 ligand.
In another embodiment, the present invention comprises painting a
patient's autologous dendritic cells with proteins, for purposes of
stimulating their
proliferation and/or differentiation. For purposes of dendritic cell
activation, any
suitable dendritic cell activator can be used including but not limited to
CD40
ligand, TRANCE, and Flt-3 ligand, as well as otherwise soluble dendritic cell
activators that retain their function when immobilized on the cell surface,
for
example, glycosyl-phosphatidylinositol-modified GM-CSF. According to other
embodiments of the present invention, any one of a number of other cell types
can
be similarly painted to enhance their proliferative or other functional
capacities,
either in vivo or ex vivo, in beneficial ways. Different cell types that can
be used
include, for example, lymphokine-activated killer cells, monocytes, B cells,
natural
killer cells, neutrophils, eosinophils, basophils, mast cells, as well as stem
and
progenitor cells, including hematopoietic stem cells, mesenchymal stem cells,
embryonic stem cells, keratinocytes, endothelial cells, islet cells,
fibroblasts,
osteoblasts, muscle cells, and neural cells. This List is not meant to be
limiting.
One-step or two-step protein transfer methods as described above are
preferred methods for expressing extrinsic proteins on the surfaces of the
present



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
18
cells for purposes of cellular auto-stimulation. Any protein that can be
produced in
a lipidated form and that therein has the capacity to incorporate into a cell
membrane (for example, a recombinant glycosyl-phosphatidylinositol-modified
protein), or that can bind to a second protein that has the capacity to
incorporate into
a cell membrane (for example, a recombinant immunoglobulin Fc-modified protein
that can bind to membrane-incorporating palmitated-protein A; or a recombinant
hisb-tagged protein that can bind to the membrane-incorporating chelator
lipid, NTA-
DTDA), is suitable to be used as a protein paint for coating the present
cells. As
used herein, the term "extrinsic protein" will refer to those fusion proteins
that are
transferred to the surface of the cell using the protein transfer methods
described
above.
The amount of protein used to paint the cells is as described above.
Gene transfer methods can also be used according to the present
invention for expressing exogenous proteins with auto-stimulatory potential on
the
surfaces of the present cells. Genetic sequences encoding these proteins can
be
introduced into cells ex vivo by any one of a number of transfection
modalities, and
these transfected cells can be administered to a patient as therapeutic cells,
according to methods well known in the art. " Genetic sequence" refers to a
polynucleotide comprising the coding sequence for a defined protein and
associated
regulatory and other non-coding sequences. Genetic sequences in the form of
cDNA clones are commercially available for a wide array of genes. Moreover,
for
those cDNA clones that are not readily accessible from commercial and other
sources, knowledge of their nucleotide sequences can be used to easily
reproduce
their cDNAs via the reverse-transcriptase polymerise chain reaction method,
incorporating the relevant gene sequences into the primers.
Transfection methods in the art encompass a host of vectors fox
delivering therapeutic genes, a host of transcriptional and translational
regulatory
elements that can be appended to the gene of interest, methods for producing
and
using these vectors, and methods for monitoring therapeutic gene efficacy and
toxicity. Significantly, inducible promoters are known in the art for
regulating the
expression of the transfected gene, so that levels of the encoded protein can
be



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
19
regulated. Those inducible promoters that can be regulated with orally-
administered
drugs are especially useful in this context. The use of transfected cells as
therapeutic
cells in this way fox the treatment of experimental osteoarthritis is
described in
Pelletier, Arthritis and Rheumatism, 40:1012-1019 (1997), wherein transfected
synovial cells were re-injected back into diseased joints. This is also
illustrated by
Yasuda, J. of Clinical Investigation, 102:1807-1814 (1998) who described the
treatment of autoimmune diabetes with transfected islet cells.
According to yet another embodiment of the present invention, gene
transfer can be used to transfer genes encoding not just a ligand, but also a
receptor
on the present cells, with the receptor chosen by virtue of its capacity to
bind to
native or extrinsic ligands on said cells.
Another significant advance provided by the present compositions and
methods is that following injection of the therapeutic cells into patients,
the
stimulatory effect mediated by the protein Iigands, transferred via coating or
gene
transfer, persist for some period of time. Where protein transfer is used to
transfer
the proteins, stimulation of the cells will eventually decrease with time, for
example,
as a consequence of dilution resulting from repeated cell divisions; in many
contexts, this progressive decrement constitutes an advantage since long-term
triggering is often not desirable. Where gene transfer is used to provide
exogenous
gene expression, expression can be maintained for prolonged periods when
suitable
expression vectors are chosen, that are well known to those familiar with the
art.
In addition, more than one protein ligand can be used to coat a single
cell. In this manner, two or more cis signaling proteins (for example,
immunoregulatory proteins) can be transferred to a cell surface. In the case
of
costimulators, combinations of such proteins can be chosen to have the
greatest
immunological effect; combinations having additive or even synergistic
benefits can
be selected and used according to the present compositions and methods. Also,
given the quantitative control afforded by protein transfer methods, where the
amount of protein that becomes transferred to the cell is determined by the
amount



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
of protein used to contact the painted cell, predetermined ratios of the
proteins to be
painted can be achieved.
Another advantage of the present methods stems from the enforced
proximity of the ligand and its receptor on the same cell surface. For certain
5 ligand:receptor pairs, for example, the costimulator B7-1 and its receptor
CD28,
there is a fast off rate, which tends to limit intercellular traps
costimulatory
signaling. By contrast, the cells of the present invention feature Iigand and
receptor
anchored to the same cell surface, which by the mass action law tends to
stabilize
the interaction between these "locked-in" binding partners, forced into
sustained
10 proximity. Thus, the present invention provides for an entirely new mode
and
kinetics of ligand:receptor signaling.
The present methods can be effected either irv vivo or ex vivo.
In ex vivo methods, the desired cells are isolated from a sample of a
patient's blood
or tissues, painted with the extrinsic protein or transfected with a gene
encoding a
15 protein of interest with auto-stimulatory potential, and then re-infused
into the
patient. Additional manipulations of the present cells axe possible during the
ex vivo
step, for example, cytokine treatment and amplification of cell numbers. IfZ
vivo
methods involve the infusion into a patient of a fusion protein that comprises
both a
homing element, that attaches the protein to the desired cell ifa vivo, and a
ligand
20 element, that triggers a receptor on the same cell in cis. Preferred homing
elements
are Fv domains of immunoglobulin proteins or cytokines with sufficient
affinity for
cognate surface receptors on the target cells. The single polypeptide chain
derivative comprising the Fv region of an immunoglobulin molecule (scFv), for
example of an immunoglobulin IgGl molecule, can also be used as a first
domain.
Specific examples of scFv include ones with specificities for cytokine and
other
receptors (for example, HER-2/neu), carcinoembryonic antigen (CEA), prostate
specific antigen (PSA), CD33 and AIRMl. HER-2/neu receptors are another
preferred target on cancer cells, since they are found on epithelial
carcinomas of the
breast and ovary. CEA is highly expressed on gastrointestinal tumors,
including
colon cancer, and immunoinhibitory properties have been attributed to it. PSA
is



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
21
highly expressed by most epithelial prostate carcinomas. CD33 and AIRM1 are
sialoadhesin family members expressed on cells of the myelomonocytic lineages.
Ligation of CD33 or AIRMI on chronic myeloid leukemia cells with antibodies
decreases cell proliferation and survival. As used herein, the term "homing
element" refers to a protein or other molecule than can bind to receptors on
specific
cells, and, when administered to a patient, attach to the cells of interst.
All of the above descriptions relating to cell type, proteiri ligands,
protein receptors, protein and gene transfer methods, and ex vivo and in vivo
and
other delivery techniques apply equally to all embodiments of the invention
disclosed herein.
The present invention is further directed to methods for determining
receptor activation thresholds and other functional properties of the present
cells.
For example, in the case of T cells, T cells can be painted with different
costimulators and their respective effects on TCR activation thresholds can be
compared. In addition, costimulator-painted T cells can be used as cellular
reagents
to evaluate the relative activities of different T cell modulatory agents. As
another
example, T cells can be painted with membrane-incorporating major
histocompatibility complex (MHC) protein:peptide antigen (or lipid antigen)
conjugates, which can trigger TCR on the same cells. In turn, these antigen
auto-
stimulated T cells can be used to test effects of extrinsic agents on TCR and
costimulator receptor activation thresholds, as well as any one of a number of
other
T cell physiological endpoints.
The present invention is further directed to methods for treating an
illness using the present cellular auto-stimulation technology. These methods
generally comprise administering to a patient an effective amount of the cells
prepared ex vivo according to the method described above, or administering
fusion
proteins that auto-stimulate cells that they home to in vivo. For the ex vivo
methods,
cells are coated with a lipidated protein or a membrane-incorporating protein
conjugate. An effective amount of these therapeutic cells are administered to
a
patient. For the irz vivo methods, one or more of said fusion proteins is



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
22
administered to a patient in an amount sufficient to result in transfer of an
effective
amount of this proteins) to an effective amount of cells.
Administration can be by any means known in the art, such as by
intravenous injection. The auto-stimulating cells as described herein can be
contained within a suitable pharmaceutical carrier for administration
according to
the present methods. "Suitable pharmaceutical carrier" is as described above.
EXAMPLES
The present examples are intended to illlustrate the invention and
IO should not be construed as limiting the invention in any way.
Example 1
The following example demonstrates a method for transferring a
B7-1 ~Fcy, fusion protein to a cell using palmitated protein A.
I5
Palmitation of protein A
Recombinant protein A (Calbiochem, La Jolla, CA) was derivatized
with the N hydroxysuccinimide ester of palmitic acid (Sigma, St. Louis, MO) as
described by Kim and Peacock, J. Inarnunol Methods, 158:57 (1993). Briefly, a
20 stock solution of the N hydroxysuccinimide ester of palmitic acid was made,
as was
a solution containing protein A in a concentration of about 1.5 mg/ml. The
solutions were mixed in a ratio of about 10 p.g ester per ml protein and
incubated at
room temperature with constant mixing for about 18 h. The lipid-derivatized
protein
A was purified as described by Huang, et al., J. Biol. Cher~a., 225:8015
(1980)
25 using a 30-ml Sephadex G-25 (Sigma) column. The protein product, referred
to
herein as "pal-pros Ao, was quantitated using a bicinchoninic acid kit (Bio-
Rad,
Richmond, CA), filter sterilized, and stored at 4° C until use.
MEnabrane incorporation of pal prot A
Daudi EL-4, JY and K562 cells (3-7 x 106/ml) were separately
30 resuspended in RPMI 1640 medium (BioWittaker, Walkersville, MD) after three



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
23
washes with this same medium. Varying concentrations of pal-prot A (or
nonderivatized protein A as negative control) were added to the cell
suspension, and
the mixture was incubated at 4° C for 2 h with constant mixing. To
assess the
incorporation of pal-prot A onto cell surfaces, cells were washed twice in
buffer
(0.25 % BSA/0.01 %o sodium azide/PBS) and then incubated on ice for 1 h with
100
p.l of 100 p.g/rnl FITC-human IgG (Sigma) diluted with the same buffer. Cells
were
washed twice in the buffer and analyzed on a FACStar~ flow cytometer (Becton
Dickinson, Mountain View, CA).
In a first set of optimization experiments, efficient incorporation of
pal-prot A was documented in four cell Lines (Fig. 1A) as detected with FITC-
conjugated human IgG. As a negative control, nonderivatized protein A lacked
the
capacity to bind to the same cells. Data from the FACStar analysis was plotted
as
arbitrary units of 1og10 fluorescence intensity versus number of EL-4 cells;
membrane incorporation was dose dependent and started to plateau at about 33
pg/mI pal-prot A, as shown in Fig. 1B. EL-4 cells were incubated with 33
p.g/ml
pal-pros A for the indicated periods of time and processed as above; pal-prot
A
incorporation was rapid, appearing immediately after addition to the cells and
reaching a plateau at ~ 1 h, as shown in Fig. 1 C. This data demonstrates that
numerous different cell lines can be used in the present protein transfer
methods,
and that the lipidated protein was incorporated into the cell fairly rapidly.
Preparation of recombinant B71 ~Fcy,
The expression plasmid pCDMB/B7Ig, encoding the complete human
B7-1 extracellular domain linked in-frame to the Fcy,, was obtained from the
American Type Culture Collection (Manassas, VA). The sequence encoding
B7-1-Fcy, was mobilized from pCDMB/B7Ig by digesting with XbaI, filling-in
with
Klenow fragment, and subsequently digesting with HindIII. The mobilized
fragment
was subcloned into the EBV episomal expression vector pREP7(3 (Invitrogen, San
Diego, CA) with HindlII and filled-in BanaHI sites. The plasmid was
transfected
into 293 cells (human kidney cell line; American Type Culture Collection), and



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
24
hygromycin B-resistant colonies were selected in serum-free UltraCulture
medium
(BioWittaker) supplemented with 10 mM glutamine, penicillin/streptomycin, and
200 ~.g/mI hygromycin B. Secreted B7-I ~Fcy, was purified from conditioned
medium by protein A-agarose (Life Technologies, Germantown, MD) affinity
chromatography and verified by SDS-PAGE. The protein was run on a 10 % SDS-
polyacrylamide gel and visualized with Coomassie blue as a dominant single
band of
--- 80 kDA under both reducing (lane 2) and nonreducing (lane 3) conditions as
shown in Fig. 2. Its identity was confirmed by ELISA, with a recombinant
protein
binding strongly to the human B7-1 specific mAb, BB-1, but not to control Ab
(data
not shown) .
B71 ~Fc~yl pj-otein transfer
Cells precoated with pal-prot A were washed once and resuspended in
RPMI 1640 medium (3-7 x 16 cells/ml). pREP7B-transfected K562 cells
(K562/REP7b) were serially incubated with 33 ~g/ml protein A for 2 h, 33
~.glml
Fcy~ fusion protein for 1 h, and BB-1 as primary Ab and FITC-conjugated goat
anti-
mouse IgG as secondary Ab. To monitor protein delivery, 106 cells were washed
twice with the same buffer as above, incubated on ice for 1 h with 1 ~.g of
human
B7-specific mAb BB-1 (PharMingen, San Diego, CA) in 100 pl of buffer. Cells
were washed once and immunostained (on ice for 1 h) with 100 ~I of I:I00
diluted
FITC-conjugated goat F(ab')2 anti-mouse Ig (Boehringer Mannheim, Indianapolis,
IN) as secondary Ab. Cells were washed once, resuspended in PBS, and analyzed
on a FACStar flow cytometer.
Figure 3A shows that when K562 cells were precoated with pal-prot
A, secondarily applied B7-1 ~Fcy~ attached to the cell surface, as detected by
,
immunostaining of the cells with anti B7-1 BB-1 mAb and FITC-conjugated goat
anti-mouse IgG. When a control Fc fusion protein (CD28 ~Fcyl) was substituted
for
B7-I ~Fcy,, no BB-I binding was observed, substantiating BB-1 mAb's B7-1
specificity. W, hen underivatized protein A was substituted for pal-prot A, no
BB-1
binding was observed, indicating the dependence of the lipid anchoring for
fusion
protein attachment.



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
Quantitatioh of exogefaously ifzcojporated B71 ~Fcy' at cell surfaces
Human B7-1 ~Fcy, was iodinated using Iodo-beads (Pierce, Rockford,
IL) according to the manufacturer's protocol, and the labeled protein was
purified
on a Sephadex G-25 column (Pharmacia, Piscataway, NJ). The specificity was
5 adjusted to 2.1 x 106cpm/p.g by addition of unlabeled B7-1 ~Fcy,. Protein
transfer
was performed as described earlier, substituting the labeled protein. All
experiments were performed in duplicate. To control .for nonspecific binding,
excess amounts of unlabeled human IgG (Sigma) were added to specifically block
the binding of B7-1 ~Fcyl to protein A. After repeated washing, counts in cell
pellets
10 were determined using a gamma counter ( 1272 Clinigamma; LKB Instruments,
Gaithersburg, MD). Counts resulting from specific binding of B7-1 ~Fcy~ were
calculated by subtracting nonspecific counts obtained with human IgG. The
average
number of molecules on a single cell was calculated according to the formula A
x B-'
x C'-' x N~, where A is the determined radioactivity (cpm) in the cell pellet,
B is the
15 specific activity of the labeled protein expressed as cpm/mol, C is the
number of
cells in the cell pellet, and NA is Avogadro's constant.
As shown in Fig. 3B, when K562 cells were precoated with excess
amounts of pal-prot A (33 pg/ml), surface levels of B7-1 ~Fcy, were dependent
on
the concentrations of applied B7-I ~Fcy,. Surface B7-1 epitope levels started
to
20 plateau at 33 p.glml, and the epitope density was similar to that on B7-1
transfected
K562 cells (data not shown). The average number of B7-1 ~Fcyl painted per cell
was
determined using 'z5I-labeled B7-I ~Fcy,. Again, K562 cells incorporated
increasing
amounts of B7-I -Fcy, as the reagent concentration was increased during the
painting
process, as shown in Table I.
25 Table I. Painti~zg of B71 ~Fcy' orzto K562 cells
B7-1 ~Fc~y, No. of B7-1 ~Fcy,lcellb .


(p,g/ml)a (mean SD)



0.033 460 240





CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
26
0.33 9,900 1,200


3.3 92,000 8,300


33 460,000 34,000


a The final concentration of B7-I ~Fcy, present during the painting procedure.
b Values were determined as described in Materials and Methods. Specific
activity of 'ZSI-labeled
B7-1 ~Fcy~ is 2.1 x 106 cpm/ug.
At the lowest concentration used (0.033 ~,g/ml), --- 460 molecules
became anchored onto each K562 cell. At the highest concentration used (33
pg/ml), about 460,000 B7-1 ~Fcy, molecules became incorporated. Taken
together,
these data establish that B7-1 ~Fcy1 can be applied to pal-prot A-coated cells
in a
quantitative fashion.
Proliferation assays
IO PBMC were isolated from fresh whole blood by Ficoll density
centrifugation. T-cells were purified by two rounds of treatment with Lympho-
kwik
(One Lambda, Canoga Park, CA). T-cell purity was verified by lack of a
proliferative response to phytohemaglutin ("PHAo) or PMA in the absence of
accessory cells. The human CD3-specific mAb HIT3a (PharMingen) was bound to
IS 96-well plates at the indicated concentrations and used in this form to
provide a first
activating signal to T-cells. Alternatively, PHA was used in soluble form as a
source of a first signal. K562 cells transfected with the negative control
vector
pREP7[3 (K562/pREP7 j3) were precoated with pal-prot A and secondarily coated
with B7-1 ~Fcy,. For each proliferation assay, I x I05 T-cells were incubated
with 4
20 x 104 B7-1 ~Fcy,-coated and mitomycin C-treated K562/REP7/3 cells for 60 h
at 37°
C. Wells were pulsed with 1 pCi [3H]thymidine for the last I6 h of the
incubation
period. Cells were harvested and counted on a Betaplate liquid scintillation
counter.
In the proliferation assays, PHA and B7-1 ~Fcy,-coated K562/REP7(3
cells (i.e., K562 cells stably transfected with the pREP7a EBV episomal
expression
25 vector) were used to provide first and second signals, respectively, to T-
cells.
K562/REP7[i cells lack detectable B7-I (data not shown) and provide a suitable
negative control fox experiments with K562/B7-1 transfected cells (i.e., K562
cells



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
27
stably transfected with a pREP7(3 vector containing human B7-1 cDNA sequence).
Surface B7-1 levels on K562/B7-1 transfected cells and B7-1 ~Fcy,-coated
K562/REP7(3 cells were determined by immunostaining, and the mean fluorescence
intensities were 550 nxn and 450 nm, respectively. As shown in Fig. 4A, in the
presence of suboptimal PHA concentrations ( < 0.5 ~ ~.g/ml), B7-1-Fcy,-coated
K562/REP7(3 cells, but not K562/REP7(3, significantly enhance T-cell
proliferation.
The costimulatory effect was comparable to that achieved with K562/B7-1
transfected cells. The B7-1 ~Fcy;lpal-prot A-dependence of the observed
costimulation was verified by showing that cells treated with a combination of
(non-
derivatized) protein A and B7-1-Fcy,, or with a combination of pal-prot A and
control CD8-Fcy,, did not enhance T-cell proliferation. In the presence of
higher
PHA concentrations ( > 1 ~,g/ml), K562/REP7(3 cells also costimulate T-cell
proliferation, although to a lesser extent than the B7-1 positive cells.
To further confirm the costimulatory function of cell-associated
B7-1 ~Fcy,, proliferation assays were performed in which plate-bound anti-
human
CD3 mAb was substituted for PHA as a more physiological first signal. In this
setting, in the presence of sub-optimal concentrations of anti-CD3 mAb ( < 10
p.g/ml) cell-associated B7-1 ~Fcyl costimulated even more effectively than
native B7-
1 expressed at equivalent levels on transfected cells, as shown in Fig. 4B.
Again,
CD8~Fcy,, used as a negative control Fc fusion protein, did not costimulate
under
the same conditions. Taken together, these results establish that B7-1 ~Fcy,,
tethered
to membranes via pal-prot A, effectively costimulates T-cell proliferation.
Effective depletion of accessory cells was documented in all T-cell
preparations by demonstrating the lack of response to PMA or PHA in the
absence
of a source for costimulation. Points shown in Figs._ 4A and B are the means
and
SEs of triplicate samples. The data are representative of at least three
independent
experiments with similar results.
Concefatration-dependerme of cell-associated B71 ~Fcy,~s costif~zulatory
activity
With an effective costimulator protein transfer method in hand,
quantitative aspects of B7-1 costimulation were evaluated. To this end, T-cell



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
28
proliferation assays were performed using K562/REP7(3 cells painted with
variable
concentrations of B7-1 ~Fcy,. The concentration dependence of B7-1 ~Fcy,-
mediated
costimulation could be readily demonstrated when a fixed suboptimal
concentration
of PHA (0.25 or 0.5 pg/ml) was used as a source of first signal, as shown in
Fig_
5A. For example, in the presence of 0.5 p.g/ml PHA, T-cell proliferation was
observed once a threshold B7-1-Fcy, concentration (0.1 p,glml) was reached,
and the
level of proliferation continued to rise with increasing B7-1-Fcy,
concentrations until
reaching a plateau at -v 3.3 p.g/ml. In the presence of a lower concentration
of PHA
(0.25 p,g/ml), T-cell proliferation was observed when a higher threshold B7-1
concentration (1 p.g/ml) was reached, indicating that costimulator thresholds
can be
modulated by the strength of the first signal.
Similar results were obtained when anti-human CD3 mAb was used
as a source of first signal instead of PHA, as shown in Fig. SB. Again, in the
presence of a fixed suboptimal concentration of plate-bound anti-CD3 mAb (0.37
or
1.1 p.g/ml), costimulation was observed only after a threshold B7-1 ~Fcy,
concentration was reached, and a further dose-dependent increase in
proliferation
was also seen. Hence, in the presence of a suboptimal first signal (whether
PHA or
anti-CD3 mAb), a threshold B7 level is required for T-cells to proliferate and
the
extent of T-cell proliferation is dictated by the costimulator level.
ELISA measuremefat of secreted cy!~kifaes and determination of a hierarchy of
B7 I
eostimulatoY thresholds for distinct cytokine responses
A total of 106 T-cells was incubated with 5 x 105 processed
K5621REP7(3 cells (B7-1 ~Fcy, positive or negative) in 4S-well plates using
either
plate-bound HIT3a or PHA as a source of first signal. Supernatants were
collected
after 48 h, and ELISAs for human IFN-y and IL-2 were performed using a
commercial ELISA kit according to manufacturer's protocol (Genzyme, Cambridge,
MA). More specifically, ELISA was used to measure T-cell cytokine secretion in
response to varying painted B7-l~Fcy, concentrations and fixed suboptimal
primary
stimulus concentrations. At a fixed PHA dose, the B7-1 ~Fcy, concentrations
eliciting minimal and maximal cytokine responses differed for IFN-y and IL-2
with



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
29
the general hierarchy being IFN-y < IL-2, as shown in Fig. 6A. A similar
hierarchy for the cytokine responses was observed when anti-CD3 mAb (3.3
~.glml)
was used as a source of first signal as shown in Fig. 6B. Fox instance, at a
B7-
1 ~Fcy, concentration of 0.33 p.glml, IFN-y output was b0 % of the maximal
response, whereas IL-2 output showed no increase above basal levels (Fig. 6B).
This observed IFN-y > IL-2 hierarchy for B7-1 costimulator thresholds matches
the
order described fox TCR activation thresholds. Having documented that B7-1
levels
can modulate the extent of T-cell proliferative responses, it was then
determined that
B7-1 levels can also dictate the quality of immune responses by altering the
ratios of
cytokines produced by activated T-cells.
Af~alysis of intracellular cytokine production and evaluation of evidence fof-
hierarchical costimulator thresholds for- cytokine respofises at the single-
cell level



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
To substantiate the ELISA findings with bulk T-cell populations,
multiparameter flow cytometric analyses were performed to assess intracellular
IFN-
y and IL-2 levels within individual cells. A total of 106 T-cells was
incubated with 5
x 105 B7-1 ~Fcy,-coated K562/REP7(3 cells in 4~-well plates for 4S h. Again,
either
5 plate-bound HIT3a or PHA was used as a source of first signal. Monesin
(Sigma)
was added to a final concentration of 3 p,M, and the mixture was incubated for
an
additional 6 h to accumulate cytokine within the cells. Cells were then
collected,
fixed by incubating them in 1fl0 p,l of fixation solution [4 %
paraformaldehyde/PBS
(pH 7.4)~ on ice for 20 min. and then washed twice with staining buffer (0.1
10 saponinl l % heat-inactivated FCS/0. I % sodium azide/Dulbecco's PBS).
Immunostaining for intracellular cytokines was performed by incubating the
cells on
ice for 1 h with 100 p.1 of the staining buffer containing 0.5 p.g of FITC-
anti-IFN-y
and 0.5 pg of PE-anti-IL-2 Abs (PharMingen). Cells were subsequently washed
once with staining buffer without saponin. T-cells were gated using forward
light
15 scatter/side light scatter parameters, and 2-5 x 104 cells were analyzed in
each run.
At low B7-1-Fcy, concentrations, the T-cell response was dominated
by IFN-y-only producers; however, at higher B7-1 ~Fcy, concentrations,
substantial
numbers of IFN-y and IL-2 double producers emerged (Fig. 7). Relatively few IL-
2
only producers were observed, even at the highest B7-1 ~Fcyl concentrations.
These
20 findings are consistent with the bulk. T-cell cytokine response data,
showing that an
IFN- (3 response requires less B7-1 costimulators than does an IL-2 response.
Example 2
The effect of temperature on membrane-incorporated protein A was
studied; the transferred protein must remain cell-bound in vivo in order to
prime T-
25 cells, which requires stable engagement of costimulators for at least
several hours.
It was determined that the reaction temperature at which a lipidated protein
is
transferred to the cell membrane has a major impact on long-term retention of
the
protein on the membrane. Protein transfer reactions were performed at
4°C, 25°C
or 37°C; palmitated protein A was transferred onto K562 cells. An hB7-1
~Fc was
30 'ZSI-labeled and transferred to the protein A-coated cells in the manner
described in



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
31
Example 1. To prevent interference of endocytosis likely to occur at
temperatures
above 4°C, the cells were treated with the metabolic inhibitors sodium
azide and 2-
deoxyglucose prior to the transfer reaction.
To determine the long-term retention of the transferred protein on the
cell membrane, the cells were thoroughly washed to remove unincorporated
proteins, and subsequently incubated in suspension fox up to three days at
37°C in
DMEM medium containing 10% fetal calf serum. ~ At several intervals, aliquots
of
the suspension were taken and cells were pelleted. The amount of radioactive
label
remaining in the cell pellet was compared to the total amount of radioactive
counts
in the aliquot. The ratio between the two was calculated as the relative
portion of
the transferred protein still retained on the cell membrane. As depicted in
Fig. ~,
there is a direct relationship between a higher protein transfer reaction
temperature
and a better long-term retention rate. More importantly, by raising the
transfer
temperature from 4°C to 37°C, the transferred proteins can
remain membrane-
bound at the physiological temperature of 37°C for three days without
significant
loss (after the initial six hours).
Example 3
C3H/HeN mice, purchased from Harlen (USA), Indianapolis, were
immunized with a cell vaccine generated from the T-50 cell line, obtained from
Avranham Hochberg, Hadassah TJniversity Hospital. The vaccine was prepared
following the procedure generally outlined in Example I, using palmitated
protein A
and mB7-1 ~Fc, m4=1BBL~Fc, and hCD40L~Fc fusion proteins. Basically, the cells
were coated with the lipidated protein A at 37°C at a ratio of 40 p.g
protein A per 40
x 10~ cells. The cells were then incubated at 4° with an equal mixture
of the three
fusion proteins at a ratio of 20 p,g total protein per 4 x 10' cells. The cell
vaccine
was injected into the mice subcutaneously at a dose of 106 cells per
injection. The
injections were given once a week and continued for three weeks. One week
after
the last injection, the animals were challenged with 106 wild-type T-50 tumor
cells,
injected intradermally on the rear flank. As Fig. 9 shows, the cell vaccine
improved
the survival rate of the immunized animals. In Fig. 9: open circle, an
untreated



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
32
control group (n=6); square, another control group that received a control
vaccine
generated by protein A transfer (n=5); closed circle, the test group that
received a
cell vaccine generated by protein transfer with immune costimulatory proteins
B7-1,
4-1BBL, and CD40L in complex with protein A (n=6).
Example 4
DBA/2J mice were purchased from The Jackson Laboratory, Maine.
The animals were inoculated intradermally with a lethal dose of L5178Y-R tumor
cells and given subcutaneous injections of a cell vaccine as a treatment on
days 5, 6,
and 7 after the tumor inoculation. The same cell vaccine in Example 3 was used
here, at a dose of 106 cells per injection. Figure 10 shows that the cell
vaccine
improved the survival rate of the treated animals. In Fig. 10: open circle, an
untreated control group (n=8); square, another control group that received a
control
vaccine generated by protein A transfer (n=8); closed circle, the test group
that
received the cell vaccine generated by protein transfer with the immune
costimulatory fusion proteins in complex with lipidate protein A (n=8).
Example 5
A vaccine was formed with palmitated protein A and FasL-Fc, B7-
1 ~Fc, 4-1BBL~Fc and CD40L~Fc fusion proteins by mixing them in vitro at three
parts of lipidated protein A and one part of each of the fusion proteins. The
protein
mixture was then injected intratumorally at 4 pg of total protein per tumor
site. The
vaccine was subsequently injected directly into a tumor; the immune
costimulatory
proteins in the vaccine modified the immunogenic property of tumor cells in
situ.
DBA mice were inoculated with a lethal dose of L5178Y-R tumor
cells. As tumor mass grew to about 50 mm2 in size, the cancer vaccine was
injected
directly into the tumor site. The vaccines were pre-assembled with palmitated
protein A, which confers the ability to anchor costimulators on the tumor
cells ija
situ according to the present methods. As shown in Fig. 11, the survival of
the mice
treated with the indirectly Iipidated costimulators was significantly
prolonged. In
Fig. 11: open circle, an untreated control group (n=6); square, another
control
group that were injected with lipidated protein A alone (n=7); closed circle,
the test



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
33
group that were injected with the immune costimulatory fusion proteins in
complex
with lipidate protein A (n=7).
Example 6
To generate a fusion protein for Fast, a coding sequence for a human
Fcy, domain, obtained from ATCC, was fused at the N-terminus of the coding
sequence for the extracellular domain of Fast following the fusion strategy
reported
in Immunity, 5:163, 1996. The purified fusion protein was fully functional, as
determined by a standard killing assay when loaded on protein A-coated cells.
Fast
is a cell surface protein that binds to another protein, Fas, found on the
surface of
other cells, for example, activated T-cells. When Fast binds to Fas, the cells
expressing Fas undergo apoptosis. Significantly, the FasL~Fc fusion protein,
after
being transferred onto the cell surface through the lipidated protein A,
retained its
apoptotic activity.
More specifically to determine whether the Fc-hFasL fusion protein
was functional after anchoring onto cell surfaces, a standard JAM assay is
performed. The effector cells were CHO cells that were painted with palmitated
protein A (pal-pros A) and subsequently with Fc fusion protein. The target
cells
were Jurkat cells that constitutively express Fas and thus are susceptible to
Fas/FasL-mediated apoptosis. A standard JAM assay was performed, according to
the protocol described by P. Matzinger (J. IfnmutZOl. Metlands, 145:185-
192,1991).
Briefly, 2 x 104 3H-thymidine-labeled target Jurkat cells were co-incubated
with 2 x
105 CHO cells (from ATCC) that were pre-coated with pal-prot A as previously
described in Example 1, and subsequently painted with 30 ~glml of Fc-hFasL
fusion
protein or control fusion protein. The cells were co-cultured in 200 ~.1 of
RPMI-10
in a 96-well plate for 18 hours at 37°C in a humidified incubator at 5
% C02. To
harvest the JAM test, the cells and their medium were aspirated onto fiber
glass
filters using a harvester (as used in Example 1 for the proliferation assays).
%
specific killing was calculated as follows: (S-E)/S x 100, where S =
spontaneous
release without effector cell, E = experimental release in the presence of
effector
cells.



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
34
The results are summarized in Figure 12. When CHO cells that were
painted with pal-prot A and Fc-hFasL were used as effector cells, significant
killing
was observed. As negative controls, when CHO cells were painted with pal-pros
A
and control Fc fusion protein (hCD80-Fc or hCD28-Fc), no specific killing was
observed. The surface anchorage-dependence of Fc-hFasL was demonstrated by the
negative control where the CHO cells were painted with unpalmitated protein A
and
Fc-FasL. The specificity of the killing was demonstrated by complete blockade
of
the killing by Fast neutralizing mAb NOK-1, whereas the control Ab did not
block
the killing.
In summary, these results demonstrate that Fc-hFasL, after being
transferred onto cell surface through pal-prot A, retains its function to
elicit
apoptosis in Fas-positive Jurkat cells.
The above examples demonstrate the efficacy of the present methods.
Through the use of lipidated proteins, fusion proteins can be transferred to
cells both
ex vivo and iyz situ. Significantly, these fusion proteins retain their
immunoregulatory function after transfer. The examples demonstrate this
retained
function against post-imitation challenge and against pre-existing tumors. The
methods were demonstrated as being effective both in vivo and in vitro.
Example 7
The following example demonstrates that murine splenic T cells
painted with a B7-1~Fcr.,:palmitated-protein A conjugate have enhanced
proliferative
potential in response to primary anti-CD3 mAb triggering of their TCR.
Me»a6rane Incorporation of B71~FcY,:palnaitated protein A and FeY,~4-
IBBL.palmitated protein A Conjugates
We applied the above methods to T cells for the first time in order to
produce pure and uniform B7-1+ or 4-1BBL+ T cells ex vivo. Murine CD4+ or
CD8+ T cells were purified from the spleens of DBA/2J mice (Jackson
Laboratory,
Bar Harbor, ME) and painted separately with palmitated-protein A and Fcy,-
derivatized B7-l, 4-1BBL, and CD28 proteins (B7-1~Fc~,,, Fc~,,~4-1BBL, and



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
CD28~Fcy,) in two sequential steps. Specifically, the purified cells were
mixed with
palmitated-protein A at 30 ~,g per 106 cells per ml in DMEM, at 37°C
for 1 h. The
cells were washed and mixed again with B7-1~Fc.~,l, Fc~,,~4-1BBL, or
CD28~Fc,~,, (as
negative control) at 30 pg per 106 cells per ml in DMEM, at 4°C fox 0.5
h. The
5 preparation of palmitated-protein A is as described above. The production of
B7-
1~Fcyl, Fc.rl~4-1BBL, and CD28~Fcr, has also been described previously (Zheng,
et
al. Cancer Res., 61:8127 (2001)).
The B7-1~FcY~-painted T cells were analyzed by flow cytometry
following immunostaining with the rat anti-mouse B7-1 mAb 1610 as primary Ab,
10 and FITC-labeled goat anti-rat conjugates as second Ab (BD Pharmingen, San
Diego, CA). Negative control rat IgG2a isotype-control mAb was also obtained
from BD Pharmingen. Of note, the rat Ab do not bind to protein A. Cells were
analyzed on a FACSAN (Becton Dckinson, Mountain View, CA), and events were
gated on live cells.
15 As shown in Figure 13A (left panels), significant B7-1-positivity was
evident on the treated cells, whether CD4+ or CD8+. In contrast, B7-1 epitopes
were undetectable on untreated splenic T cells, as well on those cells exposed
to the
B7-1 ~Fc,,, fusion protein in the absence of palmitated-protein A or in the
presence of
native (non-palmitated) protein A (data not shown).
20 To establish the generality of this T cell costimulator painting
method, Fcy,~4-1BBL was substituted for B7-1~Fc~,, in the above experimental
design. 4-1BBL-painted T cells were generated with biotin-labeled-Fcy,,~4-
IBBL,
which was subsequently detected using avidin-FITC conjugates as fluorescent
tags
(BD Pharmingen, San Diego, CA). As shown in Figure I3A (right panels),
25 significant levels of 4-1BB ligand epitopes were detectable on both CD4+
and CD8+
T cells following the painting step.
Enhafaced Proliferatiofr. of Murine Splenic T Cells paiiated with B7
I~Fc~,l:palmitated-protein A and Fcrl~4-1BBL:palmitated-protein A Conjugates



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
36
Purified murine CD4~ or CD8+ splenic T cells were prepared from
the spleens of DBA/2J purchased from the Jackson Laboratory (Bar Harbor, ME).
The cells were painted ex vivo with either B7-1~Fcy,,:palmitated-protein A or
Fc~,,~4-
1BBL:palmitated-protein A conjugates, and then plated in wells containing a
sub-
s optimal amount of plate-bound rat anti-mouse CD3 mAb (in the absence of
accessory cells). Specifically, B7-1- or 4-1BBL-painted murine T cells were
plated
in 96-well flat-bottomed plates that were pre-coated with dialyzed rat anti-
mouse
CD3 mAb (TK3; Serotec Ltd, Oxford, UK) at 1 pg per ml, 37°C for 3 h.
The cells
were seeded at 105 cells per well, in RPMI 1164 medium supplemented with 10
fetal calf serum, 15 mM HEPES, and 50 mM (3-ME. At 48 h, 3H-thymidine was
added to the culture, at 1 p,Ci per well, and the cells were harvested at 64
h. 3H-
thymidine incorporation was analyzed with a ~i-Counter.
As shown in Figure 13B, the various costimulator-painted T cells,
including B7-1- and 4-IBBL-painted CD4+ and CD8+ T cells, all proliferated
I5 vigorously (Fig. 2). In contrast, relatively little proliferation was seen
in the various
controls, including non-painted T cells treated with anti-CD3 mAb, T cells
painted
with Fc',,-derivatized murine CD28 (a non-costimulator control), T cells
treated
with native (non-lipidated) protein A in place of palmitated-protein A, and
costimulator-painted T cells in the absence of an anti-CD3 mAb stimulus.
Enhanced IL-2 production by Murirze EL-4 T Cells painted with B7
1 ~Fcr,: palrnitated protein A Conjugates
As further proof that contaminant APC in the purified T cell
preparations are not responsible for the observed proliferative response, the
homogeneous EL-4 T lymphoma line was tested. Since EL-4 cells proliferate
spontaneously in culture, IL-2 secretion was used as an alternative readout
for
stimulation. EL-4 cells were painted with B7-1~Fcy,,:palmitated-protein A
conjugates and plated in wells of 96-well flat-bottomed plates containing
decreasing
amounts of either plate-bound anti-CD3 mAb (not shown) or concanavalin A as
mitogen. Conditioned medium was taken 12 h later and analyzed for IL-2 with
the



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
37
use of a commercial ELISA kit (R&D Systems, Minneapolis, MN) as per the
manufacturer's protocols. As shown in Figure I4A, B7-I-painted EL-4 cells
secreted more IL-2, as compared to non-painted control cells under these sub-
optimal mitogen stimulation conditions.
Enhanced Proliferation of Purified Human T Cells painted with B7
1 ~Fcyr:palrnitated protein A Conjugates
In order to extend the findings beyond marine cells, human CD4~ T
cells were prepared from the peripheral blood of healthy donors. All T cells
were
purified by magnetic separation, as per the manufacturer's instructions
(Miltenyi
Biotec, Auburn, CA)_ These cells were painted with B7-1~Fcy,,:palmitated-
protein
A conjugates and assayed for proliferation under sub-optimal anti-CD3
stimulation.
Specifically, the plates were pre-coated with azide-free mouse anti-human CD3
mAb
(HIT3A; BD Pharmingen) at 0.3 pg per ml.
As shown in Figure 14B, the B7-I-painted human CD4~ T cells, like
their marine counterparts, proliferated vigorously in an anti-CD3 mAb-
dependent
manner, whereas the non-painted or control-painted cells proliferated
relatively
little.
CD28-dependence of the Enhanced Proliferation of B71 painted Marine Splenic T
cells
In order to verify that the costimulatory effect observed for B7-I-
painted marine splenic CD4+ T cells is indeed dependent upon binding to B7-1's
cognate receptor on the same T cells, CD28, we evaluated the proliferative
potential
of splenic T cells purified from CD28-~- C57BL/6 knockout mice (purchased from
the Jackson Laboratory, Bar Harbor, ME), after costimulator painting. The
cells
were processed and assayed as described above, except that the CD28-'- CD4+ T
cells were pulsed with 3H-thymidine at 12 h and harvested at 24 h.
As shown in Figure 15, the B7-1-painted, CD28-'~ knockout splenic
CD4j- T cells failed to proliferate, as compared to the robust proliferation
of wild-



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
38
type CD28-bearing cells. Signifcantly, the viability of the CD28-'- knockout
splenic
T cells was verified, by showing that they proliferated well after being
painted with
a second costimulator, 4-1BBL.
Example 8
The following example demonstrates that costimulator-painted T cells
exhibit an enhanced proliferative response to a physiologic antigenic
stimulus.
Membrane Incojporation of B71~FcY,:palrnitated protein A Conjugates on to the
surfaces of DOI1.10 TCR-transgenic T cells
DO11.10 TCR-transgenic T cells, mth OVA3zs-339 peptide antigen
specificity, generally depend on costimulator signaling from accessory cells
for their
proliferative response. We determined whether costimulator painting of these
TCR-
transgenic T cells can bypass this dependence on accessory cells for
intercellular
traps costimulation. To this end, we separately painted purified splenic T
cells and
IS the syngeneic APC cell line, PRO-IAd, with B7-1~Fcy,,:palinitated-protein A
conjugates, as described above. As shown in Figure 16A, both cell populations
exhibited substantial levels of B7-1 epitopes after this procedure.
Enhanced Proliferation of DOII.10 TCR-transgenic T cells Painted with B7
1 ~Fcy;.palmitated protein A Conjugates in Response to Specific Cognate
Antigen
The proliferative response of B7-I-painted DO11.10 TCR-transgenic
T cells (co-cultured with native PRO-IAa cells) to increasing concentrations
of the
OVAg23-339 Peptide antigen was compared to non-painted T cells from the same
source {co-cultured with PRO-IAd cells painted with B7-1~Fct,,:palmitated-
protein A
conjugates). Equal numbers of T cells were present in both cultures. As shown
in
Figure 16B, antigen-driven proliferation was markedly enhanced when B7-1 was
expressed directly on the T cells, as opposed to the accessory ceps.



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
39
Example 9
The following example demonstrates that auto-costimulation
contributes significantly to the enhanced proliferation observed in cultures
of
costimulator-painted T cells.
Distirzguislzijzg Between Cis Auto-Costimulatiozz arcd Trans Intercellular
Costirnulatiotz: Cell Mixing Experiment wztlz Mitomycin C inhibition
Within a homogeneous population of costimulator-painted,
costimulator receptor-bearing T cells, there are two possible modes of
IO costhnulation: tra>zs intercellular costimulation and cis auto-
costimulation. To
quantitatively compare the two, we devised an experimental strategy based upon
cell
mixing, as depicted in Figure 17A. A preparation of marine splenic CD4+ T
cells
was divided into two: one-half was treated with mitomycin C to render them
incapable of proliferation, whereas the other half was untreated. In turn, one-
half of
each of the mitomycin-treated and untreated cell populations was painted with
costimulator~Fc},l:palmitated-protein A conjugates.
With these four subpopulations of variably treated cells in hand, we
set up three different cell mixtures, designated A, B and C, each consisting
of equal
numbers of mitomycin C-inactivated and untreated active cells. Mixture A
consisted of costimulator-painted, inactivated cells combined with non-
painted,
active cells. Proliferation in this mixture reflects only tratzs intercellular
costimulation. By contrast, in mixture B, it is the active cells that are
costimulator-
painted, and thus, proliferation in this mixture can arise from both
intercellular trarzs
costimulation and cis-auto-costimulation. The level of cis auto-costimulation
can be
calculated by subtracting the proliferation in mixture B (trans + cis) from
that in
mixture A (cis onty).
As shown in Figure 17B, under conditions where a sub-optimal level
of anti-CD3 mAb stimulation was employed, substantially more proliferation was
observed in mixture B as compared to mixture A, for both B7-1- and 4-IBBL-
painted marine CD4''~ T cells, pointing to substantial auto-costimulation for
both



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
costimulators. Mixture C, a control in which neither the mitomycin-treated nor
the
untreated cells were painted with costimulators, provided the background level
of
costimulation-independent level of proliferation in this culture system. As
shown in
Figure 17C, a significant level of cis auto-costimulation was also observed in
an
5 analogous human CD4~ T cell co-culture system.
Distinguishirag Between Cis Auto-CostimulatiorZ and Traus Intercellular
Costimulation: Cell Mixing Experiment with CFSE-labeling
To rule out the possibility that mitomycir< C treatment somehow
10 interferes with traps intercellular costimulation, we devised an
alternative cell
mixing protocol that avoided metabolic inhibitors altogether. For this second
cell
mixing experiment, we turned to CFSE labeling, which permits quantitation of
proliferation by tracking decrements in CFSE fluorescence, which halves with
each
successive cell division. For the CFSE labeling, murine splenic CD4+ T cells
were
15 suspended at 2.5 x 106 cells per ml in PBS containing 83 nM CFDASE
(Molecular
Probes, Eugene, OR), at room temperature for 3 min. The reaction was
terminated
by adding fetal calf serum to 10%. The cells were washed twice before use in
RPMI 1164 medium supplemented with 10% fetal calf serum, IS mM HEPES, and
mM (3-ME.
20 The cell mixing scheme fox the CFSE experiment is shown in Figure
18A. In this case, mixh~re A consisted of CFSE-labeled cells combined with an
equal number of costimulator-painted, unlabeled cells, plated in wells
containing
sub-optimal levels of anti-CD3 mAb. Thus any decrement in CFSE labeling in
this
mixture reflects trams intercellular costimulation. In mixture B, it is the
CFSE-
25 labeled cells that are costimulator-painted, and thus both traps
intercellular
costimulation and cis auto-costimulation are possible. Again, as in the case
of the
mitomycin C experiment, the difference in proliferation between mixtures B
(traps
+ cis) and B (cis only) reflects the degree of cis auto-costimulation.
As shown in Figure 18B, flow cytometry at 64 h revealed
30 substantially fewer CFSE fluorescent cells in mixture B, as compared to A,
pointing



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
41
to a substantial level of cis auto-costimulation in that co-culture. As
before,
comparison of mixture A to mixture C (which lacked costimulator altogether),
pointed to a significant level of tfaps intercellular costimulation as well.
Thus, two
independent cell mixing experiments pointed to significant auto-costimulation,
in
conjunction with basal traps intercellular costimulation.
Example 10
The following example demonstrates that the relative contribution of auto
costimulation to the proliferative responses of costimulator-painted T cells
can be
augmented by diluting the T cells.
To further validate the auto-costimulation phenomenon, we
performed cell dilution analysis. We reasoned that traps intercellular
costimulation,
which is cell contact-dependent, should decrease disproportionately, as
compared to
cis auto-costimulation, when one dilutes the cultures. As shown in Figure 19,
this
was indeed the case. Specifically, human CD4+ T cells from peripheral blood
were
used to generate cell mixtures A, B and C, as described above. The mixtures
were
plated either at 3 x 105 cells per well or at half that density, 1.5 x 105
cells per well.
This two-fold dilution resulted in about a five-fold reduction in the
proliferative
activity for cell mixture A (traps), in contrast to about a 1.3-fold reduction
for cell
mixture B (traps + cis). Thus, traps, as opposed to cis, costimulation is
preferentially cell-density sensitive. This finding offers yet more evidence
that
costimulator-painted T cells can signal in both cis- and traps modes, with
their
relative dominance depending on the experimental setting.
These above examples establish the generality of the auto-
costimulated cells of the present invention (different costimulators; cells of
different
species; cells of different T cell subsets; both primary cells and cell
lines), with no
undue experimentation required to add to the series of functional auto-
costimulated
cells in this class. Moreover, the data demonstrate that the protein ligand-
painted



CA 02493590 2005-O1-25
WO 2004/011673 PCT/US2003/023039
42
cells of the present invention auto-stimulate themselves, and do so via
painted
protein ligands binding to neighboring cognate receptors on the same cell
surfaces.
Whereas particular embodiments of this invention have been
described above for purposes of illustration, it will be evident to those
skilled in the
art that numerous variations of the details of the present invention may be
made
without departing from the invention as defined in the appended claims.

Representative Drawing

Sorry, the representative drawing for patent document number 2493590 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date Unavailable
(86) PCT Filing Date 2003-07-23
(87) PCT Publication Date 2004-02-05
(85) National Entry 2005-01-25
Examination Requested 2008-07-08
Dead Application 2014-07-23

Abandonment History

Abandonment Date Reason Reinstatement Date
2005-07-25 FAILURE TO PAY APPLICATION MAINTENANCE FEE 2005-11-04
2006-07-24 FAILURE TO PAY APPLICATION MAINTENANCE FEE 2007-05-25
2013-07-23 FAILURE TO PAY APPLICATION MAINTENANCE FEE
2013-12-19 R30(2) - Failure to Respond

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $200.00 2005-01-25
Reinstatement: Failure to Pay Application Maintenance Fees $200.00 2005-11-04
Maintenance Fee - Application - New Act 2 2005-07-25 $50.00 2005-11-04
Registration of a document - section 124 $100.00 2006-01-24
Expired 2019 - Corrective payment/Section 78.6 $250.00 2006-12-12
Reinstatement: Failure to Pay Application Maintenance Fees $200.00 2007-05-25
Maintenance Fee - Application - New Act 3 2006-07-24 $100.00 2007-05-25
Maintenance Fee - Application - New Act 4 2007-07-23 $100.00 2007-05-25
Maintenance Fee - Application - New Act 5 2008-07-23 $200.00 2008-06-20
Request for Examination $800.00 2008-07-08
Maintenance Fee - Application - New Act 6 2009-07-23 $200.00 2009-06-25
Maintenance Fee - Application - New Act 7 2010-07-23 $200.00 2010-07-20
Maintenance Fee - Application - New Act 8 2011-07-25 $200.00 2011-07-22
Maintenance Fee - Application - New Act 9 2012-07-23 $200.00 2012-07-23
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
TRUSTEES OF THE UNIVERSITY OF PENNSYLVANIA
Past Owners on Record
TYKOCINSKI, MARK L.
ZHENG, GUOXING
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Abstract 2005-01-25 1 63
Claims 2005-01-25 7 261
Drawings 2005-01-25 22 326
Description 2005-01-25 42 2,266
Cover Page 2005-03-29 1 41
Claims 2011-05-05 7 194
Description 2011-05-05 42 2,287
Claims 2013-03-19 7 196
Description 2012-05-16 42 2,278
Claims 2012-05-16 7 219
Assignment 2006-01-24 7 239
PCT 2005-01-25 8 332
Assignment 2005-01-25 3 94
Correspondence 2005-03-24 1 27
Fees 2005-11-04 1 29
Prosecution-Amendment 2006-12-12 1 35
Correspondence 2006-12-18 1 18
Fees 2007-05-25 1 30
Fees 2007-05-25 1 29
Assignment 2008-02-21 4 151
Correspondence 2008-05-02 1 16
Fees 2008-06-20 1 36
Prosecution-Amendment 2008-07-08 1 33
Fees 2009-06-25 1 37
Fees 2010-07-20 1 37
Prosecution-Amendment 2010-11-25 3 135
Prosecution-Amendment 2011-05-05 15 542
Prosecution-Amendment 2011-12-14 2 65
Prosecution-Amendment 2012-05-16 13 422
Prosecution-Amendment 2012-11-21 2 69
Prosecution-Amendment 2013-03-19 12 351
Prosecution-Amendment 2013-06-19 2 84